Faculty Opinions recommendation of STAT-3-independent production of IL-17 by mouse innate-like αβ T cells controls ocular infection.

Author(s):  
Michail S Lionakis
2018 ◽  
Vol 215 (4) ◽  
pp. 1079-1090 ◽  
Author(s):  
Anthony J. St. Leger ◽  
Anna M. Hansen ◽  
Hatice Karauzum ◽  
Reiko Horai ◽  
Cheng-Rong Yu ◽  
...  

Appropriate regulation of IL-17 production in the host can mean the difference between effective control of pathogens and uncontrolled inflammation that causes tissue damage. Investigation of conventional CD4+ T cells (Th17 cells) has yielded invaluable insights into IL-17 function and its regulation. More recently, we and others reported production of IL-17 from innate αβ+ T cell populations, which was shown to occur primarily via IL-23R signaling through the transcription factor STAT-3. In our current study, we identify promyelocytic leukemia zinc finger (PLZF)–expressing iNKT, CD4−/CD8+, and CD4−/CD8− (DN) αβ+T cells, which produce IL-17 in response to TCR and IL-1 receptor ligation independently of STAT-3 signaling. Notably, this noncanonical pathway of IL-17 production may be important in mucosal defense and is by itself sufficient to control pathogenic Staphylococcus aureus infection at the ocular surface.


2001 ◽  
Vol 107 (2) ◽  
pp. 359-366 ◽  
Author(s):  
Amy L. Woodward ◽  
Jonathan M. Spergel ◽  
Harri Alenius ◽  
Emiko Mizoguchi ◽  
Atul K. Bhan ◽  
...  

1998 ◽  
Vol 63 (6) ◽  
pp. 707-714 ◽  
Author(s):  
Robert A. Collins ◽  
Dirk Werling ◽  
Sara E. Duggan ◽  
A. Patricia Bland ◽  
Keith R. Parsons ◽  
...  
Keyword(s):  
T Cells ◽  

2018 ◽  
Vol 37 (4) ◽  
pp. S221
Author(s):  
J. Hsu ◽  
A. Krishnan ◽  
S. Lee ◽  
J.M. Dodd-o ◽  
B. Kim ◽  
...  

2009 ◽  
Vol 257 (1-2) ◽  
pp. 1-4 ◽  
Author(s):  
Jyotika Sharma ◽  
Qun Li ◽  
Bibhuti B. Mishra ◽  
Judy M. Teale

2021 ◽  
Vol 9 (Suppl 1) ◽  
pp. A19.1-A19
Author(s):  
JGM Strijker ◽  
E Drent ◽  
JJF van der Hoek ◽  
R Pscheid ◽  
B Koopmans ◽  
...  

BackgroundCurrently ~50% of patients with the diagnosis of high-risk neuroblastoma will not survive due to relapsing or refractory disease. Recent innovations in immunotherapy for solid tumors are highly promising, but the low MHC-I expression of neuroblastoma represents a major challenge for T cell-mediated immunotherapy. Here, we propose a novel T cell-based immunotherapy approach for neuroblastoma, based on the use of TEG002, αβ-T cells engineered to express a defined γδ-T cell receptor, which are thought to recognize and kill target cells independent of MHC-I. In this pilot project we have tested the potential efficacy of TEG002 therapy as a novel treatment for neuroblastoma, with tumor organoids.Materials and MethodsEffector cells were created from healthy donor peripheral blood T cells. The TEG002 cells were engineered by transducing αβ-T cells with a defined Vγ9Vδ2-T cell receptor. Both the untransduced αβ-T cells and the endogenous Vγ9Vδ2-T cells from the same healthy donor were used as controls in all experiments. Activation and killing of TEG002 was tested in a co-culture setting with neuroblastoma organoids. Supernatant of the co-culture was collected at 24 hours for IFNγ ELISA to measure activation of TEG002. The dynamics of cytotoxicity were analyzed over time from 0 till 72 hours, using the live-cell imaging system IncuCyte from Sartorius®. Killing was quantified using a Caspase3/7 Green dye and the IncuCyte software. Transcriptional profiling of the neuroblastoma organoids was done by RNA sequencing and MHC-I expression of the neuroblastoma organoids was determined by flow cytometry.ResultsWe showed that 3 out of 6 neuroblastoma organoids could activate TEG002 as measured by IFNγ production. Transcriptional profiling of the neuroblastoma organoids showed that this effect correlates with an increased activity of processes involved in interferon signaling and extracellular matrix organization. Analysis of the dynamics of organoid killing by TEG002 over time confirmed that organoids which induced TEG002 activation were efficiently killed independently of their MHC-I expression. Of note, efficacy of TEG002 treatment was superior to donor-matched untransduced αβ-T cells or endogenous γδ-T cells.ConclusionsWe demonstrated that 50% of tested neuroblastoma organoids can effectively activate TEG002 and that killing of the organoids is independent of MHC-I expression. Hence, this pilot study identified TEG002 as a promising novel cellular product for immunotherapy for a subset of neuroblastoma tumors, warranting further investigations into its clinical application.Disclosure InformationJ.G.M. Strijker: None. E. Drent: A. Employment (full or part-time); Significant; Gadeta BV. J.J.F. van der Hoek: None. R. Pscheid: A. Employment (full or part-time); Significant; Gadeta BV. B. Koopmans: None. K. Ober: None. S.R. van Hooff: None. W.M. Kholosy: None. C. Coomans: A. Employment (full or part-time); Significant; Gadeta BV. A. Bisso: A. Employment (full or part-time); Significant; Gadeta BV. M. van Loenen: A. Employment (full or part-time); Significant; Gadeta BV. J.J. Molenaar: None. J. Wienke: None.


Sign in / Sign up

Export Citation Format

Share Document